Page 266 - Read Online
P. 266

Burns et al.                                                                                                                                                                        Altered filamin A enables Aβ signaling

           Aβ, the altered FLNA conformation exists not only in   kinases to hyperphosphorylate tau protein [30,32,33,52] .
           postmortem human AD brain and in triple transgenic   We know that the  linkage of altered FLNA is
           (3xTg) AD mice, but also in ICV Aβ 42 -infused wildtype   required for this Aβ signaling pathway because
           mice and in Aβ 42 -treated postmortem human control   the novel drug candidate PTI-125 prevents the
           brain [13] .                                       FLNA-α7nAChR linkage and greatly reduces tau
                                                              hyperphosphorylation [13,15] . Hyperphosphorylated
           This altered form of FLNA was evidenced by a shift   tau loses its ability to stabilize microtubules and
           in isoelectric focusing point (pI) from 5.9 in the native   dissociates from them, thereby increasing the pool
           state to 5.3 in postmortem human AD brain or brains   of free phosphorylated tau that eventually appears in
           of mouse models  [13] . An altered pI can indicate an   neurofibrillary tangles. We suggest that the signaling
           altered conformation, reflecting changes in hydrogen   of Aβ 42  via α7nAChR contributes prominently to a
           bonding, charge-charge interactions or accessibility   variety of AD-related neuropathologies in addition
           of ionizable residues within the molecule [48-50] . In   to, or perhaps elicited by, tau hyperphosphorylation
           this case, the shifted pI is resistant to complete   because these additional neuropathologies are also
           dephosphorylation by alkaline phosphatase. Hence,   reduced by PTI-125’s disruption of Aβ 42 ’s signaling
           unlike the proteopathies of tau and alpha-synuclein,   via α7nAChR [13,15] . Alternatively, they may be related
           the altered conformation of FLNA is not due to     to  the neuroinflammatory signaling  that  is also
           changes in phosphorylation state. Further studies are   disrupted by PTI-125 as discussed below. An obvious
           needed to reveal the details of FLNA’s conformational   consequence of the toxic signaling through α7nAChR
           change and whether altered FLNA is unique to AD.   is that the normal function of α7nAChR is impaired.
                                                              Because α7nAChR is an upstream regulator of the
           The induction of the altered conformation by Aβ 42  may   N-methyl-D-aspartate receptor (NMDAR) [53,54] , the
           suggest a direct interaction or some sort of cross-  impaired NMDAR signaling in AD brain and 3xTg AD
           protein templating by Aβ 42 . However, Aβ 42  and FLNA   mice is very likely related to the impaired signaling
           do not directly interact because neither protein can be   via α7nAChR. This assertion is supported by the
           co-immunoprecipitated with the other (our unpublished   improvement in signaling function of both receptors by
                                                                     [13,15]
           observations). Although it is tempting to presume   PTI-125   . Aβ 42 ’s signaling to hyperphosphorylate
           that FLNA must be in its altered conformation to   tau also contributes to eventual formation of Aβ
                                                                                                          [55,56]
           associate with α7nAChR, we hypothesize that it is   deposits and tau-containing neurofibrillary lesions  ,
           FLNA’s transmembrane recruitment to this receptor,   because disrupting this signaling via PTI-125 markedly
           induced by Aβ 42 ’s extracellular binding, that changes   reduces both Aβ deposits and neurofibrillary lesions in
                                                                                                          [13,15]
           FLNA’s  conformation.  Aβ 42  binds  α7nAChR  with   3xTg AD mice or ICV Aβ 42 -infused wildtype mice  .
           high femtomolar affinity, the highest known binding   Finally, reiterating the improved receptor function
                                                              by PTI-125 and implicating this toxic Aβ signaling
           affinity of Aβ 42 . Interestingly, prevention of the FLNA   pathway in cognitive impairment, PTI-125 also
           linkage to α7nAChR by FLNA-binding compound PTI-   improved working and spatial memory and nesting
           125 decreases Aβ 42 ’s affinity for this receptor 1,000-  behavior abnormalities in 3xTg AD mice [13] .
           10,000-fold, illustrating that FLNA enables not only
           Aβ 42 ’s toxic signaling but also its high-affinity binding
           for α7nAChR [15] . Although this observation appears   ALTERED FLNA ENABLES Aβ-INDUCED
           to suggest that altered FLNA is responsible for Aβ 42 ’s   NEUROINFLAMMATION
           binding, we propose a dynamic, sequential process:
           (1) Aβ 42  binds α7nAChR to induce FLNA recruitment;   The proteopathy of FLNA enables a second toxic
           (2) recruitment alters FLNA’s conformation; and (3)   signaling pathway of Aβ: its activation of the innate
           FLNA’s altered form secures (locks in) an ultra-high   immune receptor TLR4 [13,15] . Aβ 42  binds the CD14
           affinity Aβ 42 -α7nAChR interaction. The reasoning   co-receptor [14] , complexed with TLR4, to induce an
           behind this hypothesis is that Aβ 42  induces both the   aberrant FLNA linkage to TLR4 [13,15] , which appears
           aberrant FLNA conformation and its recruitment to   to reciprocally enable a sustained Aβ-mediated
           α7nAChR (either by ICV Aβ 42  infusion to wildtype mice   TLR4 activation [Figure 2]. Hence, the FLNA-TLR4
           or by in vitro Aβ 42  incubation of postmortem control   linkage, allowing Aβ activation of TLR4, promotes
           brain) [13,15]  and that Aβ 42  and FLNA do not themselves   persistent inflammatory cytokine release and elicits
           interact.                                          neuroinflammation characteristic of AD. As with
                                                              α7nAChR, native FLNA in control brains does not
           Enabled  by  FLNA,  Aβ 42 , in monomeric or small   associate with TLR4, unless treated with Aβ 42  by ICV
           oligomeric form [51] , signals via α7nAChR to activate   infusion to wildtype mice or by in vitro incubation of

            266                                                              Neuroimmunology and Neuroinflammation ¦ Volume 4 ¦ December 8, 2017
   261   262   263   264   265   266   267   268   269   270   271